Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters

Publication year range
1.
Blood ; 123(22): 3406-13, 2014 May 29.
Article in English | MEDLINE | ID: mdl-24615776

ABSTRACT

Idelalisib (GS-1101, CAL-101), an oral inhibitor of phosphatidylinositol 3-kinase-δ, was evaluated in a phase I study in 64 patients with relapsed indolent non-Hodgkin lymphoma (iNHL). Patients had a median (range) age of 64 (32-91) years, 34 (53%) had bulky disease (≥1 lymph nodes ≥5 cm), and 37 (58%) had refractory disease. Patients had received a median (range) of 4 (1-10) prior therapies. Eight dose regimens of idelalisib were evaluated; idelalisib was taken once or twice daily continuously at doses ranging from 50 to 350 mg. After 48 weeks, patients still benefitting (n = 19; 30%) enrolled into an extension study. Adverse events (AEs) occurring in 20% or more patients (total%/grade ≥3%) included diarrhea (36/8), fatigue (36/3), nausea (25/3), rash (25/3), pyrexia (20/3), and chills (20/0). Laboratory abnormalities included neutropenia (44/23), anemia (31/5), thrombocytopenia (25/11), and serum transaminase elevations (48/25). Twelve (19%) patients discontinued therapy due to AEs. Idelalisib induced disease regression in 46/54 (85%) of evaluable patients achieving an overall response rate of 30/64 (47%), with 1 patient having a complete response (1.6%). Median duration of response was 18.4 months, median progression-free survival was 7.6 months. Idelalisib is well tolerated and active in heavily pretreated, relapsed/refractory patients with iNHL. These trials were registered at clinicaltrials.gov as NCT00710528 and NCT01090414.


Subject(s)
Antineoplastic Agents/therapeutic use , Lymphoma, Non-Hodgkin/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Purines/therapeutic use , Quinazolinones/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Female , Humans , Lymphoma, Non-Hodgkin/diagnosis , Lymphoma, Non-Hodgkin/mortality , Male , Middle Aged , Neoplasm Grading , Purines/administration & dosage , Purines/adverse effects , Purines/pharmacokinetics , Quinazolinones/administration & dosage , Quinazolinones/adverse effects , Quinazolinones/pharmacokinetics , Salvage Therapy , Treatment Outcome
2.
Blood ; 123(22): 3390-7, 2014 May 29.
Article in English | MEDLINE | ID: mdl-24615777

ABSTRACT

In a phase 1 trial, idelalisib (GS-1101, CAL-101), a selective inhibitor of the lipid kinase PI3Kδ, was evaluated in 54 patients with relapsed/refractory chronic lymphocytic leukemia (CLL) with adverse characteristics including bulky lymphadenopathy (80%), extensive prior therapy (median 5 [range 2-14] prior regimens), treatment-refractory disease (70%), unmutated IGHV (91%), and del17p and/or TP53 mutations (24%). Patients were treated at 6 dose levels of oral idelalisib (range 50-350 mg once or twice daily) and remained on continuous therapy while deriving clinical benefit. Idelalisib-mediated inhibition of PI3Kδ led to abrogation of Akt phosphorylation in patient CLL cells and significantly reduced serum levels of CLL-related chemokines. The most commonly observed grade ≥3 adverse events were pneumonia (20%), neutropenic fever (11%), and diarrhea (6%). Idelalisib treatment resulted in nodal responses in 81% of patients. The overall response rate was 72%, with 39% of patients meeting the criteria for partial response per IWCLL 2008 and 33% meeting the recently updated criteria of PR with treatment-induced lymphocytosis.(1,2) The median progression-free survival for all patients was 15.8 months. This study demonstrates the clinical utility of inhibiting the PI3Kδ pathway with idelalisib. Our findings support the further development of idelalisib in patients with CLL. These trials were registered at clinicaltrials.gov as #NCT00710528 and #NCT01090414.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Phosphoinositide-3 Kinase Inhibitors , Purines/therapeutic use , Quinazolinones/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Purines/administration & dosage , Purines/adverse effects , Purines/pharmacokinetics , Quinazolinones/administration & dosage , Quinazolinones/adverse effects , Quinazolinones/pharmacokinetics , Recurrence , Treatment Outcome
3.
Genome Res ; 20(8): 1020-36, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20538623

ABSTRACT

Liver cytochrome P450s (P450s) play critical roles in drug metabolism, toxicology, and metabolic processes. Despite rapid progress in the understanding of these enzymes, a systematic investigation of the full spectrum of functionality of individual P450s, the interrelationship or networks connecting them, and the genetic control of each gene/enzyme is lacking. To this end, we genotyped, expression-profiled, and measured P450 activities of 466 human liver samples and applied a systems biology approach via the integration of genetics, gene expression, and enzyme activity measurements. We found that most P450s were positively correlated among themselves and were highly correlated with known regulators as well as thousands of other genes enriched for pathways relevant to the metabolism of drugs, fatty acids, amino acids, and steroids. Genome-wide association analyses between genetic polymorphisms and P450 expression or enzyme activities revealed sets of SNPs associated with P450 traits, and suggested the existence of both cis-regulation of P450 expression (especially for CYP2D6) and more complex trans-regulation of P450 activity. Several novel SNPs associated with CYP2D6 expression and enzyme activity were validated in an independent human cohort. By constructing a weighted coexpression network and a Bayesian regulatory network, we defined the human liver transcriptional network structure, uncovered subnetworks representative of the P450 regulatory system, and identified novel candidate regulatory genes, namely, EHHADH, SLC10A1, and AKR1D1. The P450 subnetworks were then validated using gene signatures responsive to ligands of known P450 regulators in mouse and rat. This systematic survey provides a comprehensive view of the functionality, genetic control, and interactions of P450s.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Gene Expression Regulation, Enzymologic , Genomics , Liver/enzymology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Child , Child, Preschool , Female , Gene Expression , Genome-Wide Association Study , Humans , Infant , Infant, Newborn , Male , Mice , Middle Aged , Pharmaceutical Preparations/metabolism , Polymorphism, Single Nucleotide , Rats , Systems Biology , Transcription, Genetic , Young Adult
4.
Blood ; 117(2): 591-4, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-20959606

ABSTRACT

Phosphatidylinositol-3-kinase p110δ serves as a central integration point for signaling from cell surface receptors known to promote malignant B-cell proliferation and survival. This provides a rationale for the development of small molecule inhibitors that selectively target p110δ as a treatment approach for patients with B-cell malignancies. We thus identified 5-fluoro-3-phenyl-2-[(S)-1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one (CAL-101), a highly selective and potent p110δ small molecule inhibitor (half-maximal effective concentration [EC(50)] = 8nM). Using tumor cell lines and primary patient samples representing multiple B-cell malignancies, we have demonstrated that constitutive phosphatidylinositol-3-kinase pathway activation is p110δ-dependent. CAL-101 blocked constitutive phosphatidylinositol-3-kinase signaling, resulting in decreased phosphorylation of Akt and other downstream effectors, an increase in poly(ADP-ribose) polymerase and caspase cleavage and an induction of apoptosis. These effects have been observed across a broad range of immature and mature B-cell malignancies, thereby providing a rationale for the ongoing clinical evaluation of CAL-101.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, B-Cell/metabolism , Lymphoma, B-Cell/metabolism , Phosphatidylinositol 3-Kinases/drug effects , Purines/pharmacology , Quinazolinones/pharmacology , Signal Transduction/drug effects , Animals , Blotting, Western , Cell Line, Tumor , Cell Separation , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors
5.
Biomarkers ; 15(3): 249-58, 2010 May.
Article in English | MEDLINE | ID: mdl-20028270

ABSTRACT

Proteomics was utilized to identify novel potential plasma biomarkers of exercise-induced muscle injury. Muscle injury was induced in nine human volunteers by eccentric upper extremity exercise. Liquid chromatography-mass spectrometry identified 30 peptides derived from nine proteins which showed significant change in abundance post-exercise. Four of these proteins, haemoglobin alpha chain, haemoglobin beta chain, alpha1-antichymotrypsin (ACT) and plasma C-1 protease inhibitor (C1 Inh), met the criterion for inclusion based on changes in at least two distinct peptides. ACT and C1 Inh peptides peaked earlier post-exercise than creatine kinase, and thus appear to provide new information on muscle response to injury.


Subject(s)
Biomarkers/blood , Exercise , Muscles/injuries , Adult , Aged , Chromatography, Liquid/methods , Cohort Studies , Complement C1 Inhibitor Protein/biosynthesis , Creatine Kinase/metabolism , Female , Hemoglobins/biosynthesis , Humans , Male , Mass Spectrometry/methods , Middle Aged , Peptides/blood , Peptides/chemistry , Proteomics/methods , alpha 1-Antichymotrypsin/blood
6.
J Appl Physiol (1985) ; 107(2): 605-12, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19498091

ABSTRACT

Muscle responses to exercise are complex and include acute responses to exercise-induced injury, as well as longer term adaptive training responses. Using Alaskan sled dogs as an experimental model, changes in muscle gene expression were analyzed to test the hypotheses that important regulatory elements of the muscle's adaptation to exercise could be identified based on the temporal pattern of gene expression. Dogs were randomly assigned to undertake a 160-km run (n=9), or to remain at rest (n=4). Biceps femoris muscle was obtained from the unexercised dogs and two dogs at each of 2, 6, and 12 h after the exercise, and from three dogs 24 h after exercise. RNA was extracted and microarray analysis used to define gene transcriptional changes. The changes in gene expression after exercise occurred in a temporal pattern. Overall, 569, 469, 316, and 223 transcripts were differentially expressed at 2, 6, 12, and 24 h postexercise, respectively, compared with unexercised dogs (based on Por=1.5). Increases in a number of known transcriptional regulators, including peroxisome proliferator-activated receptor-alpha, cAMP-responsive element modulator, and CCAAT enhancer binding protein-delta, and potential signaling molecules, including brain-derived neurotrophic factor, dermokine, and suprabasin, were observed 2 h after exercise. Biological functional analysis suggested changes in expression of genes with known functional relationships, including genes involved in muscle remodeling and growth, intermediary metabolism, and immune regulation. Sustained endurance exercise by Alaskan sled dogs induces coordinated changes in gene expression with a clear temporal pattern. RNA expression profiling has the potential to identify novel regulatory mechanisms and responses to exercise stimuli.


Subject(s)
Gene Expression Regulation , Muscle, Skeletal/metabolism , Physical Endurance/genetics , RNA, Messenger/metabolism , Snow Sports , Adaptation, Physiological/genetics , Animals , Biomarkers/blood , Biopsy , Dogs , Gene Expression Profiling/methods , Gene Regulatory Networks , Oligonucleotide Array Sequence Analysis , Time Factors
7.
PLoS Comput Biol ; 3(3): e30, 2007 Mar 02.
Article in English | MEDLINE | ID: mdl-17335344

ABSTRACT

Uncovering pathways underlying drug-induced toxicity is a fundamental objective in the field of toxicogenomics. Developing mechanism-based toxicity biomarkers requires the identification of such novel pathways and the order of their sufficiency in causing a phenotypic response. Genome-wide RNA interference (RNAi) phenotypic screening has emerged as an effective tool in unveiling the genes essential for specific cellular functions and biological activities. However, eliciting the relative contribution of and sufficiency relationships among the genes identified remains challenging. In the rodent, the most widely used animal model in preclinical studies, it is unrealistic to exhaustively examine all potential interactions by RNAi screening. Application of existing computational approaches to infer regulatory networks with biological outcomes in the rodent is limited by the requirements for a large number of targeted permutations. Therefore, we developed a two-step relay method that requires only one targeted perturbation for genome-wide de novo pathway discovery. Using expression profiles in response to small interfering RNAs (siRNAs) against the gene for peroxisome proliferator-activated receptor alpha (Ppara), our method unveiled the potential causal sufficiency order network for liver hypertrophy in the rodent. The validity of the inferred 16 causal transcripts or 15 known genes for PPARalpha-induced liver hypertrophy is supported by their ability to predict non-PPARalpha-induced liver hypertrophy with 84% sensitivity and 76% specificity. Simulation shows that the probability of achieving such predictive accuracy without the inferred causal relationship is exceedingly small (p < 0.005). Five of the most sufficient causal genes have been previously disrupted in mouse models; the resulting phenotypic changes in the liver support the inferred causal roles in liver hypertrophy. Our results demonstrate the feasibility of defining pathways mediating drug-induced toxicity from siRNA-treated expression profiles. When combined with phenotypic evaluation, our approach should help to unleash the full potential of siRNAs in systematically unveiling the molecular mechanism of biological events.


Subject(s)
Gene Silencing , Liver/metabolism , Liver/pathology , Models, Biological , PPAR alpha/metabolism , Proteome/metabolism , RNA, Small Interfering/genetics , Animals , Computer Simulation , Gene Expression Profiling/methods , Hypertrophy/chemically induced , Hypertrophy/metabolism , Liver/drug effects , Mice , PPAR alpha/genetics , RNA, Small Interfering/administration & dosage , Signal Transduction
8.
Nucleic Acids Res ; 34(16): 4486-94, 2006.
Article in English | MEDLINE | ID: mdl-16945951

ABSTRACT

RNA interference (RNAi) has great potential as a tool for studying gene function in mammals. However, the specificity and magnitude of the in vivo response to RNAi remains to be fully characterized. A molecular and phenotypic comparison of a genetic knockout mouse and the corresponding knockdown version would help clarify the utility of the RNAi approach. Here, we used hydrodynamic delivery of small interfering RNA (siRNA) to knockdown peroxisome proliferator activated receptor alpha (Ppara), a gene that is central to the regulation of fatty acid metabolism. We found that Ppara knockdown in the liver results in a transcript profile and metabolic phenotype that is comparable to those of Ppara-/- mice. Combining the profiles from mice treated with the PPARalpha agonist fenofibrate, we confirmed the specificity of the RNAi response and identified candidate genes proximal to PPARalpha regulation. Ppara knockdown animals developed hypoglycemia and hypertriglyceridemia, phenotypes observed in Ppara-/- mice. In contrast to Ppara-/- mice, fasting was not required to uncover these phenotypes. Together, these data validate the utility of the RNAi approach and suggest that siRNA can be used as a complement to classical knockout technology in gene function studies.


Subject(s)
PPAR alpha/genetics , RNA Interference , Animals , Gene Expression Profiling , Injections , Liver/metabolism , Mice , Mice, Knockout , PPAR alpha/metabolism , Phenotype , RNA, Small Interfering/administration & dosage , Transcription, Genetic
9.
Surv Ophthalmol ; 62(3): 286-301, 2017.
Article in English | MEDLINE | ID: mdl-27890620

ABSTRACT

Drugs across many pharmacologic classes induce corneal epithelial changes. Many of these drugs have cationic amphiphilic structures, with a hydrophobic ring and hydrophilic cationic amine side chain that allow them to cross cell membranes. These drugs lead to intracellular phospholipid accumulation, often manifested in the cornea by vortex keratopathy, with no effect on visual acuity and few ocular symptoms. Other drugs, notably antineoplastic agents, produce a fine diffuse corneal haze, sometimes accompanied by decreased vision that can be dose limiting. Still other medications cause crystalline epithelial precipitation that might require debridement for resolution. An understanding of the variety of drugs involved, the multiple mechanisms responsible, and the systemic diseases that produce similar changes can lead to improved management strategies for patients with corneal epithelial deposits. In most cases, drug therapy need not be modified or discontinued, but if visual acuity is affected, close collaboration with the prescribing physician can result in determining an optimized dose that treats systemic disease and minimizes these deposits. Additionally, close monitoring might be required if the drug is also associated with other ocular findings, such as optic neuropathy or retinopathy.


Subject(s)
Corneal Diseases/chemically induced , Epithelium, Corneal/drug effects , Corneal Diseases/pathology , Epithelium, Corneal/pathology , Humans
10.
Toxicol Sci ; 92(2): 578-86, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16707586

ABSTRACT

In rodents, treatment with peroxisome proliferator-activated receptor alpha (PPARalpha) agonists results in peroxisome proliferation, hepatocellular hypertrophy, and hepatomegaly. Drugs in the fibrate class of PPARalpha agonists have also been reported to produce rare skeletal muscle toxicity. Although target-driven hepatic effects of PPARalpha treatment have been extensively studied, a characterization of the transcriptional effects of this nuclear receptor/transcription factor on skeletal muscle responses has not been reported. In this study we investigated the effects of PPARalpha agonists on skeletal muscle gene transcription in rats. Further, since statins have been reported to preferentially effect type II muscle fibers, we compared PPARalpha signaling effects between type I and type II muscles. By comparing the transcriptional responses of agonists that signal through different nuclear receptors and using a selection/deselection analytical strategy based on ANOVA, we identified a PPARalpha activation signature that is evident in type I (soleus), but not type II (quadriceps femoris), skeletal muscle fibers. The fiber-type-selective nature of this response is consistent with increased fatty acid uptake and beta-oxidation, which represent the major clinical benefits of the hypolipidemic compounds used in this study, but does not reveal any obvious off-target pathways that may drive adverse effects.


Subject(s)
Muscle Fibers, Slow-Twitch/drug effects , Muscle, Skeletal/drug effects , PPAR alpha/agonists , Animals , Bezafibrate/pharmacology , Fatty Acids/metabolism , Female , Fenofibrate/pharmacology , Gene Expression Profiling , Glucose/metabolism , Hypolipidemic Agents/pharmacology , Liver/drug effects , Liver/metabolism , Muscle Fibers, Slow-Twitch/metabolism , Muscle, Skeletal/metabolism , Pyrimidines/pharmacology , Rats , Rats, Inbred Strains , Rosiglitazone , Thiazolidinediones/pharmacology , Tretinoin/pharmacology
11.
Genome Inform ; 17(2): 77-88, 2006.
Article in English | MEDLINE | ID: mdl-17503381

ABSTRACT

Toxicity is a major cause of failure in drug development. A toxicogenomic approach may provide a powerful tool for better assessing the potential toxicity of drug candidates. Several approaches have been reported for predicting hepatotoxicity based on reference compounds with well-studied toxicity mechanisms. We developed a new approach for assessing compound-induced liver injury without prior knowledge of a compound's mechanism of toxicity. Using samples from rodents treated with 49 known liver toxins and 10 compounds without known liver toxicity, we derived a hepatotoxicity score as a single quantitative measurement for assessing the degree of induced liver damage. Combining the sensitivity of the hepatotoxicity score and the power of a machine learning algorithm, we then built a model to predict compound-induced liver injury based on 212 expression profiles. As estimated in an independent data set of 54 expression profiles, the built model predicted compound-induced liver damage with 90.9% sensitivity and 88.4% specificity. Our findings illustrate the feasibility of ab initio estimation of liver toxicity based on transcriptional profiles.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Gene Expression Profiling , Liver/drug effects , Toxicogenetics/methods , Transcription, Genetic , Alanine Transaminase/blood , Algorithms , Animals , Artificial Intelligence , Aspartate Aminotransferases/blood , Bilirubin/blood , Chemistry, Clinical/methods , Cholesterol/blood , Cluster Analysis , Dose-Response Relationship, Drug , Drug-Related Side Effects and Adverse Reactions/chemically induced , Drug-Related Side Effects and Adverse Reactions/classification , Drug-Related Side Effects and Adverse Reactions/metabolism , Feasibility Studies , Liver/enzymology , Liver/pathology , Models, Biological , Pharmaceutical Preparations/classification , Rats , Rats, Sprague-Dawley , Sensitivity and Specificity
12.
Diabetes ; 52(1): 21-8, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12502489

ABSTRACT

Protein tyrosine phosphatase 1B (PTP1B) is a negative regulator of insulin receptor (IR) signal transduction and a drug target for treatment of type 2 diabetes. Using PTP1B antisense oligonucleotides (ASOs), effects of decreased PTP1B levels on insulin signaling in diabetic ob/ob mice were examined. Insulin stimulation, prior to sacrifice, resulted in no significant activation of insulin signaling pathways in livers from ob/ob mice. However, in PTP1B ASO-treated mice, in which PTP1B protein was decreased by 60% in liver, similar stimulation with insulin resulted in increased tyrosine phosphorylation of the IR and IR substrate (IRS)-1 and -2 by threefold, fourfold, and threefold, respectively. IRS-2-associated phosphatidylinositol 3-kinase activity was also increased threefold. Protein kinase B (PKB) serine phosphorylation was increased sevenfold in liver of PTP1B ASO-treated mice upon insulin stimulation, while phosphorylation of PKB substrates, glycogen synthase kinase (GSK)-3alpha and -3beta, was increased more than twofold. Peripheral insulin signaling was increased by PTP1B ASO, as evidenced by increased phosphorylation of PKB in muscle of insulin-stimulated PTP1B ASO-treated animals despite the lack of measurable effects on muscle PTP1B protein. These results indicate that reduction of PTP1B is sufficient to increase insulin-dependent metabolic signaling and improve insulin sensitivity in a diabetic animal model.


Subject(s)
Diabetes Mellitus/physiopathology , Insulin/physiology , Obesity , Protein Serine-Threonine Kinases , Protein Tyrosine Phosphatases/antagonists & inhibitors , Signal Transduction/physiology , Animals , Blood Glucose/analysis , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Insulin/blood , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Liver/enzymology , Mice , Mice, Inbred C57BL/genetics , Muscle, Skeletal/enzymology , Oligonucleotides, Antisense/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptor, Insulin/metabolism , Serine/metabolism , Tyrosine/metabolism
13.
Curr Opin Chem Biol ; 7(4): 505-10, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12941426

ABSTRACT

Toxicogenomics is the study of the structure and output of the genome as it responds to adverse xenobiotic exposure. Large-scale transcriptional analysis, made possible through microarray technologies, enables us to study and understand the complexity of the biological effects of drugs and chemicals, with the ultimate goal of separating wanted effects from adverse effects. Nuclear receptors are attractive targets for drug discovery because, as ligand-activated transcription factors, they coordinately regulate the expression of at least hundreds of genes that, in turn, control much of cellular metabolism. Through toxicogenomics, it is becoming possible to understand the therapeutic effects of agonists within the context of toxic effects, classify new chemicals as to their complete effects on biological systems, and identify environmental factors that may influence safety or efficacy of new and existing drugs.


Subject(s)
Receptors, Cytoplasmic and Nuclear/agonists , Toxicogenetics , Animals , Gene Expression Regulation , Humans , Pregnane X Receptor , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Retinoid X Receptors , Transcription Factors/genetics , Transcription Factors/metabolism , Xenobiotics/pharmacology , Xenobiotics/toxicity
14.
Mol Endocrinol ; 17(6): 1131-43, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12649327

ABSTRACT

Phosphorylation of stress-activated kinase p38, a MAPK family member, was increased in liver of ob/ob diabetic mice relative to lean littermates. Treatment of ob/ob mice with protein tyrosine phosphatase 1B (PTP1B) antisense oligonucleotides (ASO) reduced phosphorylation of p38 in liver-to below lean littermate levels-and normalized plasma glucose while reducing plasma insulin. Phosphorylation of ERK, but not JNK, was also decreased in ASO-treated mice. PTP1B ASO decreased TNFalpha protein levels and phosphorylation of the transcription factor cAMP response element binding protein (CREB) in liver, both of which can occur through decreased phosphorylation of p38 and both of which have been implicated in insulin resistance or hyperglycemia. Decreased p38 phosphorylation was not directly due to decreased phosphorylation of the kinases that normally phosphorylate p38-MKK3 and MKK6. Additionally, p38 phosphorylation was not enhanced in liver upon insulin stimulation of ASO-treated ob/ob mice (despite increased activation of other signaling molecules) corroborating that p38 is not directly affected via the insulin receptor. Instead, decreased phosphorylation of p38 may be due to increased expression of MAPK phosphatases, particularly the p38/ERK phosphatase PAC1 (phosphatase of activated cells). This study demonstrates that reduction of PTP1B protein using ASO reduces activation of p38 and its substrates TNFalpha and CREB in liver of diabetic mice, which correlates with decreased hyperglycemia and hyperinsulinemia.


Subject(s)
Diabetes Mellitus/metabolism , Liver/metabolism , Mitogen-Activated Protein Kinases/metabolism , Obesity , Oligonucleotides, Antisense/metabolism , Protein Tyrosine Phosphatases/metabolism , Animals , Blood Glucose/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Dual Specificity Phosphatase 2 , Insulin/metabolism , JNK Mitogen-Activated Protein Kinases , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Phosphatase 2 , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Random Allocation , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases
15.
Mol Cancer Ther ; 2(2): 151-63, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12589032

ABSTRACT

Acetylation of histones in chromatin is one mechanism involved in the regulation of gene transcription and is tightly controlled by the balance of acetyltransferase and deacetylase (HDAC) activities. In cancer, some genes are repressed by the inappropriate recruitment of HDACs, e.g., tumor suppressor genes. To understand the genomic effects of HDAC inhibition on gene transcription we studied the gene expression profiles of T24 bladder and MDA breast carcinoma cells treated with three HDAC inhibitors, suberoylanilide hydroxamic acid, trichostatin A, and MS-27-275. The gene expression profiles of the HDAC inhibitors were generally similar to one another and differed substantially from those produced by structurally related inactive analogues; consequently, the changes in gene expression are mechanism-based. Hierarchical clustering of expression profiles demonstrated a greater similarity between the two hydroxamate-containing inhibitors (suberoylanilide hydroxamic acid and trichostatin A) than with MS-27-275. This difference was also supported by cell phenotypic experiments. As many genes were down-regulated as up-regulated by HDAC inhibitor treatment. Comparison of the data sets defined a common ("core") set of 13 genes regulated by all of the HDAC inhibitors in three cell lines, 8 up-regulated and 5 down-regulated. Ten of 13 genes were confirmed in dose response studies in T24 cells by quantitative-PCR. The core regulated genes are involved predominantly in cell cycle/apoptosis and DNA synthesis in response to HDAC inhibitors. These data will aide in understanding the complex set of events in cells in response to chromatin remodeling induced by HDAC inhibition, which may be responsible for antitumor effects.


Subject(s)
Breast Neoplasms/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors , Urinary Bladder Neoplasms/genetics , Benzamides/pharmacology , Breast Neoplasms/enzymology , Chromatin/chemistry , DNA Primers/chemistry , DNA, Neoplasm/analysis , Gene Expression Profiling , Humans , Hydroxamic Acids/pharmacology , Oligonucleotide Array Sequence Analysis , Pyridines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Tumor Cells, Cultured , Up-Regulation , Urinary Bladder Neoplasms/enzymology
16.
Environ Health Perspect ; 112(4): 423-7, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15033591

ABSTRACT

DNA microarrays and related tools offer promise for identification of pathways involved in toxic responses to xenobiotics. To be useful for risk assessment, experimental data must be challenged for reliability and interlaboratory reproducibility. Toward this goal, the Hepatotoxicity Working Group of the International Life Sciences Institute (ILSI) Health and Environmental Sciences Institute (HESI) Technical Committee on Application of Genomics to Mechanism-Based Risk Assessment evaluated and compared biological and gene expression responses in rats exposed to two model hepatotoxins--clofibrate and methapyrilene. This collaborative effort provided an unprecedented opportunity for the working group to evaluate and compare multiple biological, genomic, and toxicological parameters across different laboratories and microarray platforms. Many of the results from this collaboration are presented in accompanying articles in this mini-monograph, whereas others have been published previously. (Italic)In vivo(/Italic) studies for both compounds were conducted in two laboratories using a standard experimental protocol, and RNA samples were distributed to 16 laboratories for analysis on six microarray platforms. Histopathology, clinical chemistry, and organ weight changes were consistent with reported effects. Gene expression results demonstrated reasonable agreement between laboratories and across platforms. Discrepancies in expression profiles of some individual genes were largely due to platform differences and approaches to data analysis rather than to biological or interlaboratory variability. Despite these discrepancies there was overall agreement in the biological pathways affected by these compounds, demonstrating that transcriptional profiling is reproducible between laboratories and can reliably identify affected pathways necessary to provide mechanistic insight. This effort represents an important first step toward the use of transcriptional profiling in risk assessment.


Subject(s)
Gene Expression Profiling , Liver/drug effects , Liver/pathology , Oligonucleotide Array Sequence Analysis , Animals , Anti-Allergic Agents/toxicity , Clofibrate/toxicity , Data Collection , Hypolipidemic Agents/toxicity , Male , Methapyrilene/toxicity , Observer Variation , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Risk Assessment
17.
Environ Health Perspect ; 112(4): 439-48, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15033593

ABSTRACT

Several studies using microarrays have shown that changes in gene expression provide information about the mechanism of toxicity induced by xenobiotic agents. Nevertheless, the issue of whether gene expression profiles are reproducible across different laboratories remains to be determined. To address this question, several members of the Hepatotoxicity Working Group of the International Life Sciences Institute Health and Environmental Sciences Institute evaluated the liver gene expression profiles of rats treated with methapyrilene (MP). Animals were treated at one facility, and RNA was distributed to five different sites for gene expression analysis. A preliminary evaluation of the number of modulated genes uncovered striking differences between the five different sites. However, additional data analysis demonstrated that these differences had an effect on the absolute gene expression results but not on the outcome of the study. For all users, unsupervised algorithms showed that gene expression allows the distinction of the high dose of MP from controls and low dose. In addition, the use of a supervised analysis method (support vector machines) made it possible to correctly classify samples. In conclusion, the results show that, despite some variability, robust gene expression changes were consistent between sites. In addition, key expression changes related to the mechanism of MP-induced hepatotoxicity were identified. These results provide critical information regarding the consistency of microarray results across different laboratories and shed light on the strengths and limitations of expression profiling in drug safety analysis.


Subject(s)
Anti-Allergic Agents/toxicity , Gene Expression Regulation , Liver/drug effects , Liver/pathology , Methapyrilene/toxicity , Oligonucleotide Array Sequence Analysis , Animals , Male , Observer Variation , RNA/analysis , Rats , Rats, Sprague-Dawley , Reproducibility of Results
18.
Mol Cell Endocrinol ; 203(1-2): 155-68, 2003 May 30.
Article in English | MEDLINE | ID: mdl-12782412

ABSTRACT

Protein tyrosine phosphatases are important regulators of insulin signal transduction. Our studies have shown that in insulin resistant and diabetic ob/ob and db/db mice, reducing the levels of protein tyrosine phosphatase 1B (PTP1B) protein by treatment with a PTP1B antisense oligonucleotide resulted in improved insulin sensitivity and normalized plasma glucose levels. The mechanism by which PTP1B inhibition improves insulin sensitivity is not fully understood. We have used microarray analysis to compare gene expression changes in adipose tissue, liver and muscle of PTP1B antisense-treated ob/ob mice. Our results show that treatment with PTP1B antisense resulted in the downregulation of genes involved in lipogenesis in both fat and liver, and a downregulation of genes involved in adipocyte differentiation in fat, suggesting that PTP1B antisense acts through a different mechanism than thiazolidinedione (TZD) treatment. In summary, microarray results suggest that reduction of PTP1B may alleviate hyperglycemia and enhance insulin sensitivity by a different mechanism than TZD treatment.


Subject(s)
Adipose Tissue/metabolism , Gene Expression Regulation , Lipids/biosynthesis , Liver/metabolism , Oligonucleotides, Antisense/pharmacology , Protein Tyrosine Phosphatases/physiology , Adipose Tissue/cytology , Animals , Blood Glucose/drug effects , Cell Differentiation/drug effects , Down-Regulation/drug effects , Gene Expression Profiling , Gene Expression Regulation/drug effects , Insulin Resistance , Mice , Mice, Obese , Muscles/metabolism , Oligonucleotides, Antisense/therapeutic use , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/antagonists & inhibitors
19.
Environ Health Perspect ; 112(4): 428-38, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15033592

ABSTRACT

Microarrays have the potential to significantly impact our ability to identify toxic hazards by the identification of mechanistically relevant markers of toxicity. To be useful for risk assessment, however, microarray data must be challenged to determine reliability and interlaboratory reproducibility. As part of a series of studies conducted by the International Life Sciences Institute Health and Environmental Science Institute Technical Committee on the Application of Genomics to Mechanism-Based Risk Assessment, the biological response in rats to the hepatotoxin clofibrate was investigated. Animals were treated with high (250 mg/kg/day) or low (25 mg/kg/day) doses for 1, 3, or 7 days in two laboratories. Clinical chemistry parameters were measured, livers removed for histopathological assessment, and gene expression analysis was conducted using cDNA arrays. Expression changes in genes involved in fatty acid metabolism (e.g., acyl-CoA oxidase), cell proliferation (e.g., topoisomerase II-Alpha), and fatty acid oxidation (e.g., cytochrome P450 4A1), consistent with the mechanism of clofibrate hepatotoxicity, were detected. Observed differences in gene expression levels correlated with the level of biological response induced in the two in vivo studies. Generally, there was a high level of concordance between the gene expression profiles generated from pooled and individual RNA samples. Quantitative real-time polymerase chain reaction was used to confirm modulations for a number of peroxisome proliferator marker genes. Though the results indicate some variability in the quantitative nature of the microarray data, this appears due largely to differences in experimental and data analysis procedures used within each laboratory. In summary, this study demonstrates the potential for gene expression profiling to identify toxic hazards by the identification of mechanistically relevant markers of toxicity.


Subject(s)
Clofibrate/toxicity , Gene Expression Profiling , Hypolipidemic Agents/toxicity , Liver/drug effects , Liver/pathology , Oligonucleotide Array Sequence Analysis , Animals , Male , Observer Variation , Polymerase Chain Reaction , RNA/analysis , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Risk Assessment
20.
Toxicology ; 181-182: 537-50, 2002 Dec 27.
Article in English | MEDLINE | ID: mdl-12505364

ABSTRACT

Microarray analysis of gene expression has become a powerful approach for exploring the biological effects of drugs and other chemicals. In toxicology research, gene expression profiling may help identify hazards by comparing results for an experimental compound with a database, and establish mechanistic hypotheses through examination of discrete gene changes. Here we examine the hepatic effects of a thienopyridine inhibitor of NF-kappa B-mediated expression of cellular adhesion proteins. In a 3-day toxicity study in Sprague-Dawley rats, A-277249 induced hypertrophy of the liver and elevated serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST) and alkaline phosphatase (ALP). To investigate mechanism, microarray analysis was done on RNA from livers of A-277249-treated rats. Gene expression profiles from A-277249 were compared with a database of profiles from fifteen known hepatotoxins. Agglomerative hierarchical cluster analysis showed A-277249 to have a profile most similar to the aromatic hydrocarbons Aroclor 1254 and 3-methylcholanthrene (3MC), two known activators of the aryl hydrocarbon nuclear receptor (AhR). Several genes regulated by the AhR, including cytochrome P450 1A1, were upregulated by A-277249. In addition, several genes involved in apoptosis and cell cycle were differentially expressed consistent with cell turnover, hypertrophy and hyperplasia observed by histology. Results from this study indicate that A-277249 hepatic toxicity is mediated by the AhR either directly or through effects on NF-kappa B, and demonstrate the utility of microarray analysis for the rapid identification of toxic hazards for new chemical entities.


Subject(s)
Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/biosynthesis , Oligonucleotide Array Sequence Analysis/methods , Receptors, Aryl Hydrocarbon/physiology , Signal Transduction/drug effects , Toxicology/instrumentation , Animals , Blotting, Western , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Gene Expression/drug effects , Liver/pathology , Liver Function Tests , NF-kappa B/genetics , NF-kappa B/metabolism , Proteins/metabolism , Pyridines/toxicity , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptors, Aryl Hydrocarbon/drug effects , Receptors, Aryl Hydrocarbon/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL